GOT1 Human, Active

Glutamic-Oxaloacetic Transaminase 1 Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

GOT1 Human Recombinant E.coli produced in E.Coli is a single, non-glycosylated polypeptide chain containing 433 amino acids (1-413 a.a.) and having a molecular mass of 48.4 kDa. The GOT1 is fused to a 20 amino acid His-Tag at N-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Glutamate oxaloacetate transaminase 1 (GOT1), also known as aspartate aminotransferase 1 (AST1), is an enzyme involved in amino acid metabolism. GOT1 has two isoforms, GOT1 and GOT2, which are found in the cytoplasm and mitochondria respectively. Both isoforms are homodimers and share high homology. They play a crucial role in the urea cycle and the tricarboxylic acid cycle.
Description
This product consists of the human recombinant GOT1 enzyme, which has been expressed in E. coli and purified to a high degree. The protein sequence includes amino acids 1 to 413 of the GOT1 protein with an additional 20 amino acid His-Tag fused at the N-terminus. The molecular weight of the recombinant protein is 48.4 kDa. Glycosylation is not present in this recombinant protein.
Physical Appearance
The product is a clear and colorless solution that has been sterilized by filtration.
Formulation
The GOT1 protein is supplied in a solution at a concentration of 0.5mg/ml. The solution is buffered with 20mM Tris-HCl at pH 8.0 and contains 2mM DTT, 0.1M NaCl, and 20% glycerol.
Stability
To ensure product stability, store the vial at 4°C if it will be used within 2-4 weeks. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein such as 0.1% HSA or BSA is advised for long-term storage to maintain protein stability. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of GOT1 Human Recombinant is determined to be greater than 95.0% using SDS-PAGE analysis.
Biological Activity
The specific activity of the enzyme is determined to be greater than 50 units/mg. One unit of activity is defined as the amount of enzyme required to catalyze the conversion of 1 μmole of α-ketoglutarate to L-Glutamate per minute at a pH of 8.0 and a temperature of 25°C.
Synonyms
EC 2.6.1.1, Aspartate aminotransferase 1, Transaminase A, GIG18.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAPPSVFAEV PQAQPVLVFK LTADFREDPD PRKVNLGVGA YRTDDCHPWV LPVVKKVEQK IANDNSLNHE YLPILGLAEF RSCASRLALG DDSPALKEKR VGGVQSLGGT GALRIGADFL ARWYNGTNNK NTPVYVSSPT WENHNAVFSA AGFKDIRSYR YWDAEKRGLD LQGFLNDLEN APEFSIVVLH ACAHNPTGID PTPEQWKQIA SVMKHRFLFP FFDSAYQGFA SGNLERDAWA IRYFVSEGFE FFCAQSFSKN FGLYNERVGN LTVVGKEPES ILQVLSQMEK IVRITWSNPP AQGARIVAST LSNPELFEEW TGNVKTMADR ILTMRSELRA RLEALKTPGT WNHITDQIGM FSFTGLNPKQ VEYLVNEKHI YLLPSGRINV SGLTTKNLDY VATSIHEAVT KIQ.

Q&A

What is human GOT1 and what is its primary function in cellular metabolism?

GOT1 (Glutamate Oxaloacetate Transaminase 1) is a cytosolic enzyme that catalyzes the reversible transfer of an amino group from L-aspartate to α-ketoglutarate, forming oxaloacetate and L-glutamate. This reaction is a critical component of the malate-aspartate shuttle (MAS), which facilitates the transfer of reducing equivalents (NADH) from the cytosol to the mitochondria. The MAS is essential for maintaining redox balance between cellular compartments, as NADH cannot directly cross the mitochondrial membrane.

In metabolic terms, GOT1 functions as a key node connecting amino acid metabolism with central carbon metabolism. The enzyme plays critical roles in:

  • Transferring reducing equivalents between cellular compartments

  • Supporting mitochondrial oxidative phosphorylation

  • Regulating cellular aspartate and glutamate levels

  • Mediating metabolic adaptations to various physiological states

How is GOT1 gene expression regulated in different tissues?

GOT1 expression exhibits tissue-specific regulation with particular responsiveness to environmental and physiological stimuli. In brown adipose tissue (BAT), GOT1 is markedly induced by cold exposure through a well-characterized signaling cascade:

  • Cold stress activates the sympathetic nervous system, releasing norepinephrine

  • Norepinephrine binds to β-adrenergic receptors (βAR) on brown adipocytes

  • This activates the βAR-cAMP-PKA signaling pathway

  • Downstream, PGC-1α and its shorter isoform NT-PGC-1α are activated

  • PGC-1α/NT-PGC-1α bind to the GOT1 gene promoter at estrogen-related receptor (ERR) binding motifs

  • This binding increases GOT1 transcription and ultimately protein levels

This regulatory pathway appears to be highly specific for GOT1, as other components of the malate-aspartate shuttle (including GOT2, MDH1, and MDH2) remain unchanged during cold exposure .

What distinguishes GOT1 activity from other transaminases in experimental settings?

When designing experiments to study GOT1, researchers must consider several distinguishing characteristics:

  • Subcellular localization: Unlike GOT2 (mitochondrial), GOT1 is primarily cytosolic, requiring different fractionation techniques for isolation

  • Substrate specificity: While GOT1 primarily catalyzes the aspartate-oxaloacetate reaction, it exhibits varying degrees of activity with other amino acid substrates

  • Inhibitor sensitivity: GOT1 can be selectively inhibited by aminooxyacetate (AOA), which is often used experimentally to distinguish GOT1-dependent effects

  • Regulatory mechanisms: GOT1 activity is subject to allosteric regulation and post-translational modifications distinct from other transaminases

For accurate measurement, researchers typically use spectrophotometric assays that monitor NADH oxidation coupled to malate dehydrogenase activity, or direct measurement of oxaloacetate or aspartate levels using mass spectrometry or chromatography techniques .

How does GOT1 contribute to the malate-aspartate shuttle and cellular redox balance?

The malate-aspartate shuttle (MAS) serves as a critical pathway for transferring reducing equivalents from cytosolic NADH to the mitochondrial electron transport chain. GOT1 occupies a central position in this shuttle with the following mechanistic steps:

  • In the cytosol, malate dehydrogenase (MDH1) converts oxaloacetate to malate while oxidizing NADH to NAD+

  • GOT1 regenerates oxaloacetate by transferring an amino group from aspartate to α-ketoglutarate

  • Malate and α-ketoglutarate enter the mitochondria via specific transporters

  • Inside mitochondria, malate is converted back to oxaloacetate by MDH2, generating NADH

  • Mitochondrial GOT2 completes the cycle, transferring the amino group from glutamate to oxaloacetate

Through this cycle, GOT1 facilitates the regeneration of cytosolic NAD+, which is essential for sustaining glycolysis and other NAD+-dependent processes. Importantly, the shuttle effectively transfers the reducing equivalents of cytosolic NADH to the mitochondria without physically moving the NADH molecule itself .

Experimental evidence demonstrates that overexpression of GOT1 activates the MAS, as shown by:

  • Decreased cytosolic NADH levels (measured using the Peredox NADH sensor)

  • Increased mitochondrial respiration

  • Enhanced production of deuterium-labeled malate when cells are cultured with [4-2H]-glucose

What is the relationship between GOT1 activity and cellular fuel preference?

GOT1 plays a crucial role in determining cellular fuel preference, particularly in tissues like brown adipose tissue (BAT). Research findings demonstrate that:

  • GOT1 overexpression in brown adipocytes enhances fatty acid oxidation (FAO) while reducing glucose oxidation

  • Conversely, GOT1 knockout shifts fuel preference from fatty acids toward glucose

The mechanism behind this metabolic shift involves several interconnected processes:

  • Enhanced MAS activity increases the transfer of reducing equivalents to mitochondria

  • This supports mitochondrial respiration and oxidative metabolism

  • GOT1-mediated MAS activation stimulates AMPK through changes in the ADP/ATP ratio

  • Activated AMPK phosphorylates and inhibits acetyl-CoA carboxylase (ACC)

  • Inhibited ACC reduces malonyl-CoA production, relieving inhibition of carnitine palmitoyltransferase 1 (CPT1)

  • This ultimately promotes fatty acid transport into mitochondria and subsequent oxidation

Paradoxically, GOT1 overexpression also increases glucose uptake and glycolysis while reducing glucose oxidation. This is likely because enhanced MAS activity supports the recycling of cytosolic NAD+, which is required for sustained glycolytic flux .

How does GOT1 contribute to cellular adaptation during hypoxia?

GOT1 plays a critical role in cellular adaptation to hypoxic conditions through several mechanisms:

  • Aspartate homeostasis: GOT1 activity influences intracellular aspartate pools, which become limited during oxygen restriction. GOT1 knockout cells show increased steady-state levels of aspartate.

  • Glycolytic support: GOT1 is required for the hypoxia-induced upregulation of glycolysis. When GOT1 is knocked out, glucose uptake and lactate production (key indicators of glycolytic activity) are significantly reduced during hypoxia.

  • Redox balance maintenance: GOT1 contributes to cytoplasmic NAD+ regeneration, which becomes critical when oxygen availability limits the electron transport chain's ability to reoxidize NADH.

Experimental evidence shows that the hypoxia-induced metabolic shift to increased glycolysis is significantly attenuated in GOT1-knockout cells. Importantly, this phenotype can be reversed by ectopic expression of GOT1, confirming that GOT1 enzymatic activity, rather than just aspartate concentration, is required for proper hypoxic adaptation .

What are the optimal methods for measuring GOT1 enzyme activity?

Accurate measurement of GOT1 enzyme activity requires careful consideration of assay conditions and potential interference from other enzymes. The following methodological approaches are recommended:

Spectrophotometric coupled assay:

  • Principle: The GOT1 reaction is coupled to malate dehydrogenase, which oxidizes NADH to NAD+

  • Detection: Decrease in absorbance at 340 nm (NADH absorption maximum)

  • Advantages: Continuous, real-time measurement; widely accessible equipment

  • Limitations: Potential interference from other NADH-consuming reactions

Direct measurement of reaction products:

  • Methods: High-performance liquid chromatography (HPLC) or mass spectrometry

  • Measurement: Quantification of oxaloacetate, aspartate, glutamate, or α-ketoglutarate

  • Advantages: Direct measurement without coupling reactions; higher specificity

  • Limitations: Requires specialized equipment; not real-time

Isotope tracing approaches:

  • Using deuterium-labeled glucose ([4-2H]-glucose) to track GOT1-dependent flux through the malate-aspartate shuttle

  • Measuring labeled metabolites (e.g., [M+1]-malate) using mass spectrometry

  • This approach can reveal dynamic aspects of GOT1 function in intact cellular systems

For optimal results, researchers should perform activity assays on purified enzyme preparations or carefully prepared subcellular fractions to distinguish cytosolic GOT1 from mitochondrial GOT2 activity .

How can researchers develop effective GOT1 knockout or overexpression models?

Creating reliable GOT1 genetic models is essential for studying its function. Based on published methodologies, the following approaches have proven effective:

For GOT1 knockout models:

  • CRISPR/Cas9-mediated gene editing targeting the Got1 gene exons

  • Cre-loxP conditional knockout systems for tissue-specific deletion

  • Validation through Western blotting, enzyme activity assays, and metabolite profiling

For GOT1 overexpression models:

  • Generation of knock-in mice using a CAG-loxP-neo-3xpA(stop)-loxP-Got1 construct inserted into the Rosa26 locus

  • Use of Cre-mediated recombination to excise the stop cassette, enabling tissue-specific Got1 expression

  • Isolation of stromal vascular fraction (SVF) from BAT of transgenic mice, followed by Cre-mediated recombination and differentiation into brown adipocytes

Critical validation steps include:

  • Confirming GOT1 protein levels by immunoblotting

  • Measuring enzyme activity in cytosolic fractions

  • Assessing MAS activity using NADH sensor systems (e.g., Peredox)

  • Verifying expected metabolic phenotypes (e.g., changes in aspartate levels, glycolysis, fatty acid oxidation)

What isotope tracing strategies are most informative for studying GOT1-mediated metabolic fluxes?

Isotope tracing provides critical insights into metabolic pathway activity and is particularly valuable for studying GOT1-dependent processes. The following approaches have proven informative:

[4-2H]-glucose tracing:

  • This substrate transfers deuterium to NADH during glycolysis

  • The labeled NADH ([4-2H]-NADH) then transfers deuterium to metabolites via various dehydrogenases

  • In the context of the malate-aspartate shuttle, [4-2H]-NADH transfers deuterium to oxaloacetate via MDH1, generating [2-2H]-malate

  • Quantification of labeled malate provides a direct measure of flux through the MAS

13C-glutamine or 13C-glucose tracing:

  • These tracers allow researchers to track carbon flow through central metabolic pathways

  • Analysis of labeled aspartate, malate, and TCA cycle intermediates reveals GOT1's contribution to various metabolic routes

  • Comparing labeling patterns between wild-type and GOT1-modified cells can uncover metabolic rewiring

15N-glutamate or 15N-aspartate tracing:

  • These tracers specifically track nitrogen transfer reactions catalyzed by transaminases

  • Useful for distinguishing GOT1 activity from other metabolic pathways

Analysis typically employs liquid chromatography-mass spectrometry (LC-MS) to quantify labeled metabolites. The labeling patterns and kinetics provide insights into pathway activity and flux directionality that cannot be obtained from steady-state measurements alone .

What is the therapeutic potential of recombinant GOT1 (rGOT1) in cerebral ischemia?

Recombinant GOT1 (rGOT1) has shown promising neuroprotective effects in cerebral ischemia models through a glutamate-scavenging mechanism:

Mechanism of action:

  • During ischemic stroke, excessive glutamate accumulates in brain tissue and serum

  • Administered rGOT1, together with its co-substrate oxaloacetate, catalyzes the conversion of glutamate to α-ketoglutarate and aspartate

  • This reduces extracellular glutamate concentration, thereby limiting excitotoxicity

  • The blood-brain barrier impairment during ischemia facilitates the glutamate-scavenging effect

Experimental evidence:

  • In rat models of middle cerebral artery occlusion (MCAO), rGOT1 administration significantly reduced serum glutamate levels

  • MR spectroscopy demonstrated decreased brain glutamate levels following rGOT1 treatment

  • rGOT1 administration provided significant neuroprotection, reducing infarct volume and improving functional outcomes

  • The therapeutic window appears to extend up to at least 1 hour after reperfusion

Dosing considerations:

  • Effective doses in animal models increased serum GOT activity at least two to three-fold

  • Observational studies in stroke patients suggest that individuals with naturally higher GOT activity (average 17 U/l vs. 11 U/l) had better neurological outcomes

  • The therapeutic effect requires co-administration of oxaloacetate to drive the GOT1 reaction toward glutamate consumption

How does GOT1 function change in hypoxic conditions and what are the metabolic consequences?

GOT1 plays a critical role in cellular adaptation to hypoxia with significant metabolic implications:

Changes in GOT1 function during hypoxia:

  • GOT1 activity becomes crucial for maintaining metabolic homeostasis when oxygen is limited

  • GOT1 supports cytoplasmic NAD+ regeneration, which is essential when the electron transport chain is limited by oxygen availability

  • GOT1 contributes to aspartate homeostasis, which is typically depleted during hypoxia

Metabolic consequences of GOT1 deletion in hypoxic conditions:

  • Impaired glycolytic response: GOT1 knockout cells show significantly reduced glucose uptake and lactate production during hypoxia

  • Altered aspartate metabolism: GOT1 knockout leads to increased steady-state aspartate levels, which persist during hypoxia

  • Compromised hypoxic adaptation: Cells lacking GOT1 show impaired ability to adapt their metabolism to oxygen limitation

Importantly, the metabolic defects in GOT1 knockout cells can be rescued by ectopic expression of GOT1, confirming that the enzymatic activity of GOT1, rather than simply aspartate levels, is required for proper hypoxic adaptation .

What is the role of GOT1 in brown adipose tissue thermogenesis?

GOT1 plays a previously unrecognized but critical role in brown adipose tissue (BAT) thermogenesis and cold adaptation:

Cold-induced GOT1 regulation:

  • GOT1 is dramatically induced by cold exposure in BAT while other MAS components remain unchanged

  • This induction occurs via β-adrenergic receptor signaling through a PKA-PGC-1α pathway

  • Cold exposure increases GOT1 from barely detectable levels to significant expression in BAT

Functional impact on thermogenesis:

  • Enhanced fatty acid oxidation: GOT1 activation promotes preferential utilization of fatty acids as fuel for thermogenesis

  • Metabolic rewiring: GOT1 overexpression increases glucose uptake and glycolysis while paradoxically reducing glucose oxidation

  • Uncoupled respiration: GOT1-mediated MAS activation supports increased UCP1-dependent uncoupled respiration

Consequences of GOT1 deletion:

  • BAT-specific Got1 knockout mice show decreased capacity for fatty acid oxidation

  • These mice compensate by shifting fuel preference toward glucose

  • The metabolic rewiring in Got1 knockout brown adipocytes involves increased pyruvate utilization

This research positions GOT1 as a cold-inducible metabolic switch that activates the malate-aspartate shuttle to support BAT's preference for fatty acids as thermogenic fuel during cold exposure .

How can researchers leverage GOT1 to manipulate cellular metabolic pathways?

GOT1 represents a powerful node for metabolic manipulation due to its central position connecting multiple pathways. Researchers can leverage GOT1 in several sophisticated ways:

Metabolic flux redirection:

  • Modulating GOT1 activity can shift the balance between fatty acid oxidation and glucose oxidation

  • This approach can be useful for studying metabolic flexibility and substrate preference

  • By combining GOT1 manipulation with isotope tracing, researchers can quantify how metabolic fluxes are redistributed

Redox balance manipulation:

  • GOT1 overexpression enhances cytosolic NAD+ regeneration via MAS activation

  • This can be exploited to study how NAD+/NADH ratios influence various cellular processes

  • Particularly valuable for research on glycolysis-dependent cellular functions

Mitochondrial function modulation:

  • GOT1-mediated MAS activation increases the supply of reducing equivalents to mitochondria

  • This provides a tool for modulating mitochondrial respiration without directly targeting ETC complexes

  • Can be used to study respiratory capacity and substrate utilization

Methodological approaches:

  • Inducible expression systems for temporal control of GOT1 activity

  • Chemical inhibitors like aminooxyacetate (AOA) for acute inhibition

  • Subcellular targeting of GOT1 to specific compartments to study localized metabolic effects

  • Combination with metabolomics and fluxomics for comprehensive pathway analysis

What are the current challenges in translating GOT1-focused research to clinical applications?

Translating GOT1-focused research to clinical applications faces several significant challenges:

Dosing and pharmacokinetic considerations:

  • Effective doses in animal models require increasing systemic GOT activity two to three-fold

  • Determining equivalent effective doses in humans remains challenging

  • The co-substrate oxaloacetate may require high doses that could have potential side effects

Therapeutic window limitations:

  • For applications like ischemic stroke, GOT1-based treatments appear effective when administered within 1-2 hours after onset

  • This relatively narrow window requires rapid intervention systems

  • Further research is needed to determine if the window can be extended

Delivery and formulation challenges:

  • Recombinant proteins like rGOT1 face stability and immunogenicity issues

  • Optimal formulation with co-substrates like oxaloacetate needs further development

  • Targeted delivery to specific tissues remains difficult

Regulatory and translational hurdles:

  • Moving from pre-clinical studies to human trials requires extensive safety data

  • The glutamate-scavenging approach differs mechanistically from traditional drug approaches

  • Demonstrating superiority over existing treatments is necessary

Knowledge gaps:

  • Better understanding of GOT1's role in various pathological conditions is still needed

  • The long-term consequences of manipulating GOT1 activity remain unclear

  • Additional mechanistic studies are required to identify potential off-target effects

How do alterations in GOT1 activity affect mitochondrial function and cellular stress responses?

GOT1 activity has profound effects on mitochondrial function and cellular stress responses through several interconnected mechanisms:

Effects on mitochondrial function:

  • Respiratory capacity: GOT1-mediated MAS activation increases the supply of reducing equivalents to the mitochondrial electron transport chain, enhancing respiratory capacity

  • Fuel utilization: GOT1 overexpression promotes fatty acid oxidation while reducing glucose oxidation in mitochondria

  • Uncoupled respiration: In brown adipocytes, GOT1 supports UCP1-dependent uncoupled respiration and thermogenesis

Impact on cellular stress responses:

  • AMPK activation: Enhanced mitochondrial uncoupling associated with GOT1 overexpression activates AMPK by increasing the ADP/ATP ratio

  • Hypoxic adaptation: GOT1 is critical for proper metabolic adaptation to oxygen limitation, supporting glycolytic flux when oxidative phosphorylation is compromised

  • Redox homeostasis: By influencing NAD+/NADH ratios, GOT1 affects cellular redox status, which is crucial for antioxidant defense systems

Metabolic rewiring:

  • GOT1 knockout cells compensate for metabolic deficiencies through alternative pathways

  • For example, GOT1-deficient brown adipocytes shift toward increased pyruvate utilization

  • This metabolic plasticity represents an important cellular stress response mechanism

Understanding these effects provides valuable insights for research on mitochondrial diseases, metabolic disorders, and conditions involving cellular stress responses such as ischemia-reperfusion injury and cancer metabolism .

Comparison of Metabolic Parameters Between Wild-type and GOT1-modified Systems

Metabolic ParameterGOT1 OverexpressionGOT1 KnockoutReference
Cytosolic NADHDecreasedIncreased
Aspartate levelsDecreasedIncreased
Glucose uptakeIncreasedDecreased
Lactate productionIncreasedDecreased
Fatty acid oxidationEnhancedReduced
Glucose oxidationReducedEnhanced
AMPK activationIncreasedDecreased
Mitochondrial respirationEnhancedReduced

Therapeutic Parameters for rGOT1 in Cerebral Ischemia Model

ParameterValue/FindingNotesReference
Effective dose12.88 mg/kg (80 U/kg)In combination with oxaloacetate (3.5 mg/kg)
Therapeutic windowUp to 1h after reperfusionMay extend to 2h after ischemia onset
GOT activity increase2-3 foldRequired for therapeutic effect
Serum glutamate reductionSignificantKey mechanism of neuroprotection
Brain glutamate reductionSignificantMeasured by MR spectroscopy
Infarct volume reductionSignificantCompared to controls
Functional improvementSignificantBehavioral assessment

Key experimental controls for GOT1 functional studies

When designing experiments to study GOT1 function, researchers should implement the following essential controls:

  • Enzyme specificity controls:

    • Use aminooxyacetate (AOA) as a specific inhibitor to confirm GOT1-dependent effects

    • Include parallel experiments with GOT2 manipulation to distinguish isoform-specific functions

    • Employ catalytically inactive GOT1 mutants to separate enzymatic from non-enzymatic functions

  • Metabolite rescue experiments:

    • Supplement aspartate to determine if phenotypes are due to aspartate depletion

    • Add cell-permeable α-ketoglutarate analogs to bypass GOT1 in relevant pathways

    • Test if malate/aspartate supplementation can rescue MAS deficiency

  • Subcellular fractionation quality control:

    • Verify purity of cytosolic fractions using markers like GAPDH

    • Confirm absence of mitochondrial contamination using markers like COX IV

    • Validate activity measurements with recombinant GOT1 standards

  • Genetic model validation:

    • Confirm complete knockout/knockdown at protein level, not just mRNA

    • Verify rescue of phenotypes with wild-type but not catalytically inactive GOT1

    • Check for compensatory changes in related enzymes (GOT2, MDH1, MDH2)

Optimizing isotope tracing experiments for GOT1-dependent pathways

Isotope tracing experiments require careful design to maximize information about GOT1-dependent metabolic fluxes:

  • Tracer selection considerations:

    • Use [4-2H]-glucose to specifically trace NADH-dependent reactions in the MAS

    • For TCA cycle connections, [U-13C]-glutamine provides more direct labeling of α-ketoglutarate

    • [1,2-13C]-glucose can trace pyruvate entry into the TCA cycle via PDH versus PC

  • Time course optimization:

    • Short time points (2.5-10 minutes) capture fast isotopic equilibration in the MAS

    • Intermediate time points (30-60 minutes) reveal compensatory metabolic adaptations

    • Longer measurements (2-24 hours) show stable isotope incorporation patterns

  • Sample preparation:

    • Rapid quenching is essential to prevent post-collection metabolism

    • Cold methanol extraction preserves metabolite integrity

    • Careful separation of extracellular and intracellular fractions

  • Data analysis approaches:

    • Isotopologue distribution analysis reveals pathway contributions

    • Flux estimation requires mathematical modeling of tracer dynamics

    • Integration with transcriptomic and proteomic data enhances interpretation

  • Advanced applications:

    • Pulse-chase designs can distinguish pool sizes from flux rates

    • Multi-tracer experiments provide complementary pathway information

    • In vivo tracing in GOT1 transgenic models connects to physiological relevance

Emerging questions about GOT1's role beyond canonical functions

Recent discoveries have opened new avenues for GOT1 research beyond its classical role in the malate-aspartate shuttle:

  • Signaling functions:

    • Is GOT1 involved in amino acid sensing pathways?

    • Does GOT1 participate in retrograde signaling from cytosol to nucleus?

    • Can GOT1 activity modulate cellular response to stress independent of its metabolic role?

  • Tissue-specific functions:

    • How does GOT1 function differ in metabolically distinct tissues?

    • Are there tissue-specific post-translational modifications that regulate GOT1?

    • Do tissue-specific binding partners alter GOT1 function?

  • Non-enzymatic roles:

    • Does GOT1 have protein-protein interaction functions independent of its enzymatic activity?

    • Could GOT1 serve as a metabolite carrier or buffer in certain cellular contexts?

    • Are there moonlighting functions of GOT1 in different subcellular locations?

  • Evolutionary considerations:

    • How has GOT1 function evolved across species with different metabolic needs?

    • Do alternative GOT1 isoforms serve specialized functions?

    • What can comparative studies of GOT1 across species reveal about its fundamental roles?

Integrating GOT1 research with emerging technologies

Advancing GOT1 research will benefit from integration with cutting-edge technologies:

  • Single-cell metabolomics:

    • Analyze GOT1-dependent metabolic heterogeneity at the single-cell level

    • Identify cell-specific roles of GOT1 in complex tissues

    • Correlate GOT1 activity with cell state and fate decisions

  • CRISPR screening approaches:

    • Perform genome-wide CRISPR screens to identify synthetic lethal interactions with GOT1

    • Use CRISPR activation/inhibition to map genetic networks connected to GOT1 function

    • Develop CRISPR base editing to create specific GOT1 variants

  • Spatial metabolomics:

    • Map GOT1 activity and metabolite distributions within tissues

    • Correlate GOT1 function with microenvironmental factors

    • Understand compartmentalized metabolism in complex systems

  • Protein structure and dynamics:

    • Apply cryo-EM to study GOT1 in complex with binding partners

    • Use hydrogen-deuterium exchange mass spectrometry to map conformational changes

    • Develop specific small molecule modulators based on structural insights

  • Systems biology approaches:

    • Integrate multi-omics data to position GOT1 in comprehensive metabolic networks

    • Develop computational models of GOT1-dependent metabolic fluxes

    • Apply machine learning to predict context-dependent functions of GOT1

Product Science Overview

Structure and Function

GOT1 is a homodimeric enzyme, meaning it consists of two identical subunits. It is involved in the biosynthesis of L-glutamate from L-aspartate or L-cysteine . This process is essential for maintaining the levels of glutamate, which is the major excitatory neurotransmitter in the vertebrate central nervous system . Additionally, GOT1 acts as a scavenger of glutamate in brain neuroprotection .

The enzyme’s activity is also significant in hepatic glucose synthesis during development and in adipocyte glyceroneogenesis . Using L-cysteine as a substrate, GOT1 regulates the levels of mercaptopyruvate, an important source of hydrogen sulfide .

Clinical Significance

GOT1 is overexpressed in many cancers, making it a potential therapeutic target . Its role in regulating cell proliferation by participating in amino acid metabolism, especially glutamine metabolism, has been highlighted in several studies . This makes GOT1 a critical enzyme in understanding and potentially treating various cancers.

Diagnostic Use

The enzyme is also known as Serum Glutamic-Oxaloacetic Transaminase (SGOT) or Aspartate Aminotransferase (AST). It is commonly measured in blood tests to assess liver health . Elevated levels of SGOT/AST can indicate liver damage or other medical conditions .

Recombinant GOT1

Recombinant GOT1 refers to the enzyme produced through recombinant DNA technology, which allows for the production of large quantities of the enzyme for research and therapeutic purposes. This recombinant form retains the same activity and properties as the naturally occurring enzyme, making it valuable for various applications in biochemistry and medicine.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.